WO2001016601A1 - Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites - Google Patents

Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites Download PDF

Info

Publication number
WO2001016601A1
WO2001016601A1 PCT/US2000/023985 US0023985W WO0116601A1 WO 2001016601 A1 WO2001016601 A1 WO 2001016601A1 US 0023985 W US0023985 W US 0023985W WO 0116601 A1 WO0116601 A1 WO 0116601A1
Authority
WO
WIPO (PCT)
Prior art keywords
nmda
nrl
nr2a
subunits
receptor
Prior art date
Application number
PCT/US2000/023985
Other languages
English (en)
Inventor
David H. Farb
Shelley Russek
Ming-Kuei Jang
Terrell Gibbs
Nader Yaghoubi
Original Assignee
Trustees Of Boston University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22540293&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2001016601(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Trustees Of Boston University filed Critical Trustees Of Boston University
Priority to EP00959706A priority Critical patent/EP1212618B1/fr
Priority to DE60041773T priority patent/DE60041773D1/de
Priority to AU70981/00A priority patent/AU782250B2/en
Priority to CA002383047A priority patent/CA2383047A1/fr
Publication of WO2001016601A1 publication Critical patent/WO2001016601A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • G01N33/9406Neurotransmitters
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J1/00Normal steroids containing carbon, hydrogen, halogen or oxygen, not substituted in position 17 beta by a carbon atom, e.g. estrane, androstane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J3/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by one carbon atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70571Receptors; Cell surface antigens; Cell surface determinants for neuromediators, e.g. serotonin receptor, dopamine receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • the NMDA receptor subtype is known to have a fundamentally important role in CNS function. Ongoing work from many laboratories has established the involvement of NMDA receptors in multiple aspects of brain development, synaptic plasticity associated with long-term potentiation, and pathology related to glutamate-mediated excitotoxicity . In particular, neuropathological mechanisms mediated by NMDA receptors have recently been implicated in neurological disorders including ischemic stroke, kindling epileptogenesis, and schizophrenia .
  • the NMDA receptor is composed of two different types of subunits, the NR1 and the NR2. The NR1 subunit is ubiquitously expressed throughout the CNS, particularly in the cerebral cortex, hippocampus, and olfactory bulb. While homomeric NMDA receptors composed of NR1 subunits are activated by NMDA, heteromeric receptors composed of both NR1 and NR2 subunits exhibit greater responses to NMDA.
  • Alternative splicing of three exons, a , ⁇ , and ⁇ (also referred to in the art as Nl, CI , and C2 , respectively) generates eight isoforms of the mRNA encoding the NR1 protein.
  • a schematic of the NR1 splice variants is shown in Fig. 1 in which the presence or absence of any of the three alternatively spliced exons is designated by a subscript within the name.
  • Figure 2 lists the eight splice variants and the alternatively spliced exons present in each variant. Exons, ⁇ , ⁇ , and ⁇ , code for 21, 37, and 38 amino acid sequences, respectively.
  • exon 5 which corresponds to exon 5
  • ⁇ and ⁇ exons corresponding to exons 21 and 22, respectively
  • a stop codon is contained within the ⁇ exon. Its removal by alternative splicing leads to the inclusion of a new 22 amino acid sequence .
  • any one or a combination of the three exons imparts differential pharmacological properties to the NMDA receptor.
  • inclusion of the Nl exon leads to a decrease in agonist affinity, but an increase in current amplitude.
  • a number of consensus PKC phosphorylation sites have been identified within the CI exon; however, a functional role for these sites has not been fully elucidated.
  • Other differences imparted by alternative splicing include sensitivity to potentiation by spermine, Zn 2+ , and activators of PKC.
  • NRl splice variants An examination of the levels of NRl splice variants in the developing rat brain has revealed regional differences in the pattern of expression.
  • This divergent pattern in the expression of the NRl splice variants may provide a degree of functional diversity in NMDA receptor function that underlies the regional heterogeneity in certain NMDA receptor-dependent processes, including synaptic consolidation, potentiation, and plasticity.
  • NR2A The four subtypes of the NR2 subunit: NR2A, NR2B, NR2C, and NR2D were identified through molecular cloning studies. Studies indicate that the inclusion of different NR2 subtypes within the heteromeric NMDA receptor alters the pharmacological properties of NMDA receptor function providing another level of functional diversity to the receptor. For example, differences in Ca 2+ permeability, sensitivity to Mg 2+ block, glycine potentiation, and offset kinetics can be attributed to the presence of specific NR2 subunits in the heteromeric receptor.
  • the NR2 subunits have about 50% sequence homology between members and have approximately 15% sequence homology with the NRl subunits. They are incapable of forming functional NMDA-activiated channels following homomeric expression, but rather serve to potentiate the NMDA response and modify pharmacological properties when coexpressed with NRl subunits. Recent work has shown that the NR2A and NR2B subunits are tyrosine- phosphorylated, whereas the NRl subunits are not.
  • the NR2 subunits exhibit a region-specific and temporal - specific pattern of expression.
  • the NR2B subunit is mainly expressed in the rat forebrain, whereas the NR2C subunit is found predominantly in the cerebellum. Differences in the expression of the NR2 subunits may be responsible for imparting functional diversity to NMDA receptor function from one cell type to the next .
  • the present invention relates to a method for identifying a subunit specific modulator of the N-methyl- D-aspartate (NMDA) receptor.
  • the method involves providing a plurality of NMDA receptors which differ in their subunit identity.
  • the receptors are contacted with a neurotransmitter recognition site ligand in the' presence and absence of a candidate modulator.
  • Receptor activity is then assayed, with an increase or decrease in activity in at least one, but not all members of the plurality of NMDA receptors, in the presence but not the absence of a candidate modulator, being an indication that the candidate modulator is a subunit specific modulator.
  • the subunit identity of the subset of the NMDA receptors to determine the subunit specificity of the candidate modulator.
  • Various combinations of NMDA receptor subunits are provided.
  • Figure 1 is a schematic of the proposed structure of the alternative splice variants of the NMDARl (NRl) mRNA.
  • Figure 2 is a compilation of graphical representations of data which indicate that PS inhibits
  • FIGS 1(A) through 1(D) are representative traces showing the inhibitory effect of 100 ⁇ M PS on kainate- induced currents of oocytes injected with (A) rat brain poly (A) + RNA, (B) GluRl cRNA, (C) GluR3 cRNA, (D) GluR6 cRNA.
  • the kainate concentration used in (A) - (C) was 100 ⁇ M, and in (D) was 10 ⁇ M.
  • the solid bar represents the period of kainate
  • FIG. 1(E) is a graph of relative current for the indicated Kainate concentration.
  • the administration of PS (open symbols) is seen to decreases maximum kainate responses of GluRl (•, O) , GluR3 ( ⁇ , D), and GluR ⁇ (A,
  • GluR6 (A) receptors Results are expressed as percentage change in the peak 100 ⁇ M (GluRl and GluR3) or 10 ⁇ M (GluR6) kainate- induced current in the presence of PS. Each data point is the mean of three experiments; error bars indicate S.E.M.
  • Figure 3 is a compilation of graphical representations of data which indicate that neuroactive steroids modulate NMDA responses of oocytes injected with specific NMDA receptor subunits.
  • Figure 2(A) indicates the potentiation of the 100 ⁇ M NMDA response by PS in oocytes injected with NR1 100 + NR2A cRNA. The solid bar indicates the period of NMDA exposure; the open bar indicates the period of PS exposure.
  • Figure 2(B) indicates inhibition of the 100 ⁇ M NMDA response by 3 ⁇ 5/3S in oocytes injected with NR1 100 + NR2A cRNA. The solid bar indicates the period of NMDA exposure; the shaded bar indicates the period of 3 ⁇ ;5j8S exposure.
  • Figure 2(C) indicates modulation of agonist efficacy by PS and 3 ⁇ 5 ⁇ S in oocytes injected with NR1 100 + NR2A cRNA.
  • PS 100 ⁇ M
  • 3 ⁇ 5/3S (100 ⁇ M) markedly reduces the NMDA J ma ⁇ with little effect on EC 5Q .
  • Peak NMDA responses are normalized to the peak 100 ⁇ M NMDA response.
  • Each data point represents the mean of three experiments. Error bars represent standard error.
  • Figure 2 (D) is a graph indicating the concentration dependence of steroid modulation of the NMDA response of oocytes injected with NR1 100 + NR2A cRNA.
  • NMDA 100 ⁇ M
  • concentration of PS 100
  • 3/35/3S
  • 3 5 ⁇ S 3 5 ⁇ S
  • the peak NMDA- induced current is expressed relative to the average of control NMDA responses determined before application of steroid and after steroid washout. Points indicate mean of 6 (PS and 3o;5/3S) , and 4 (3/35/3S) , experiments. Error jbars indicate S.E.M.
  • NMDA 300 ⁇ M
  • concentration of steroid 100 ⁇ M
  • the peak NMDA-induced current is expressed relative to the average of control NMDA responses determined before application of steroid and after steroid washout. Points indicate mean of 6 (PS), 3 (3/35/3S) , and 3 (3c*5/3S) experiments. Error bars indicate S.E.M. Smooth curves are derived from fits to the Michaelis-Menten equation, as fits to the logistic equation yielded Hill coefficients close to 1, with no significant improvement in sum of squares (F-test, P > 0.05).
  • Figure 4 is a graphical representation of data indicating that 3o;5
  • Figure 5 is a bar graph of data indicating inhibition of NMDA- induced neuronal death by 3o;5/3S isomers is stereospecific .
  • the neuronal death caused by acute exposure to 30 ⁇ M NMDA is nearly abolished by 5/3 isomers.
  • 3 ⁇ 5 ⁇ S 100 ⁇ M
  • 100 ⁇ M 3(S5 ⁇ S potentiates neuronal death.
  • Results are expressed as mean % neuronal death ⁇ S.E.M., with the number of experiments indicated in parentheses.
  • * Indicates a statistically significant (P ⁇ 0.05) difference from NMDA control; t indicates a statistically significant difference from 3/35/3S and 3 ⁇ 5/3S .
  • Figure 6 is a graphical representation of data which indicates that modulation of NMDA- induced current by steroid sulfates and hemisuccinates .
  • A Inhibition of the 100 ⁇ M NMDA- induced current by 3ce5/3HS in a Xenopus oocyte expressing NR1 100 and NR2A subunits.
  • B Inhibition of the NMDA- induced current by 3o;5/5S is shown for comparison.
  • C Potentiation of the 100 ⁇ M NMDA- induced current by PHS in a Xenopus oocyte expressing NR1 100 and NR2A subunits.
  • D Potentiation of the NMDA- induced current by PS is shown for comparison.
  • Figure 7 indicates inhibition of NMDA-mediated neuronal death by pregnane steroids is dependent upon the C3 ester group.
  • A Structures of 3 ⁇ 5/?, 3 ⁇ 50S, 3Q;5J8F, 3 ⁇ 5jSHO, 3 ⁇ 50HS, 3 ⁇ 5/3HG. Note that 3 ⁇ 5/3 and 3 x5 ⁇ F are uncharged, while the other three are negatively charged.
  • B Negatively charged pregnane steroids are more effective as inhibitors of the NMDA response. Bars show mean percentage decrease in the 100 ⁇ M NMDA- induced current in the presence of 100 ⁇ M of the indicated steroid in oocytes expressing NR1 100 and NR2A subunits. Error bars indicate S.E.M.
  • Figure 8 indicates pregn-5-ene steroid-mediated exacerbation of NMDA receptor function is dependent upon the C3 ester group.
  • A Structures of pregnenolone (P) , PS, PF, PHO, PHS, and PHG. Note that pregnenolone and PF are uncharged, while the other three are negatively charged.
  • B PHS and PHG show greatest potentiation of the NMDA response . Bars show mean percentage increase in the 100 ⁇ M NMDA-induced current in the presence of 100 ⁇ M of the indicated steroid in oocytes expressing NR1 100 and NR2A subunits. Error bars indicate S.E.M. Number of oocytes is given in parentheses. * Significant (p ⁇
  • Figure 9 is a graphical representation of data indicating steroid modulation of acute NMDA- induced neuronal death is through an interaction with the NMDA receptor.
  • 5/3 steroid isomers with A-B ring junction in the cis conformation
  • a negatively charged group at C3 such as 3 ⁇ 5/3s and 3/35/3S
  • C3 stereochemistry is not a determinant of activity.
  • the more planar 5 ⁇ isomers (with the A-B ring junction in the trans conformation) are active positive modulators only when the negatively charged group at C3 is in the ⁇ orientation; 3/35 ⁇ S and PS potentiate the NMDA response, while 3 ⁇ 5 ⁇ !S is inactive or possesses much reduced inhibitory activity.
  • Figure 11 is a representation of current recordings obtained from an oocytes bathed in solutions containing either Ba 2+ or Ca + ions.
  • the current responses were induced by co-application of 10 mM glycine and 80 mM NMDA (single bars) . Double bars indicate the co-application of 100 mM PS in addition to NMDA and glycine.
  • Figure 12 contains graphical representations of PS dose-responses for NR1/NR2A receptors. Presented are normalized current responses obtained from oocytes injected with (A) : NR1 000 /NR2A, NR1 100 /NR2A; (B) : NR1 001 /NR2A, NR1 101 /NR2A; (C) : NR1 Q11 /NR2A, NRl ⁇ /NR2A mRNAs .
  • Data points are averaged normalized peak NMDA-induced current responses obtained from oocytes injected with (A) NR1 011 /2A or (B) NRl ⁇ n /2A mRNAs. Concentration-response data for NMDA (circles) and for
  • Figure 16 contains two graphs of data indicating 3 ⁇ 5/3S proportionately modulates NR1 Q11 /2A and NRl ⁇ /2A splice variants at pH 7.5. Data points are averaged normalized peak NMDA-induced current responses obtained from oocytes injected with (A) NR1 Q11 /2A or (B) NRl ⁇ /2A mRNAs. Concentration-response data for NMDA (circles) and for NMDA + 100 ⁇ M PS (squares) were obtained in the presence of 10 ⁇ M glycine.
  • Figure 17 contains bar graphs of data indicating neuroactive steroid modulation of NR1 Q11 /2A and NRl 1 /2A splice variants at pH 7.5.
  • Bars indicate (A) the percentage potentiation of the maximum NMDA response by 100 ⁇ M PS for NR1 Q11 /2A (50%) and on NRl ⁇ n /2A (103%; *p ⁇ 5xl0 "7 ) receptors and (B) the percentage inhibition of the maximum NMDA response by 100 ⁇ M 3cx5 ⁇ S for NR1 Q11 /2A (56%) and NRl n ⁇ /2A (59%; p>0.05) receptors.
  • Dose- response data (from the same oocytes as in Figures 15 and 16) were individually fitted to the logistic equation to determine the maximum NMDA response. Bars depict the average potentiation/inhibition of the maximum NMDA response for the number of oocytes given in parentheses. Error bars represent S.E.M.
  • Figure 18 is a bar graph of data indicating PS induced modulation is NRl ⁇ splice variant dependent.
  • Figure 19 contains two graphs of data indicating pregnenolone sulfate (PS) differentially modulate NR1 011 /2A and NRl n ⁇ /2A splice variants at pH 8.5. Data points are averaged normalized peak NMDA-induced current responses obtained from oocytes injected with (A) NR1 011 /2A or (B) NRl ⁇ n /2A mRNAs. Concentration-response data for NMDA (circles) and for NMDA + 100 ⁇ M PS
  • Figure 20 contains bar graphs of data which indicate PS differentially induced modulation of NR1 011 /2A and NRl ⁇ /2A splice variants at pH 7.5 and 8.5. Bars indicate (A) the percentage potentiation of the maximum NMDA response by 100 ⁇ M PS for NR1 011 /2A (50%) and on
  • Dose-response data (from the same oocytes as in Figures 15 and 19) were individually fitted to the logistic equation to determine the maximum NMDA response. Bars depict the average potentiation of NMDA response for the number of oocytes given in parantheses .
  • Error bars represent S.E.M
  • Figure 21 is a graphical representation of data indicating effect of pH on PS induced modulation of NR1 011 /2A and NRl ⁇ /2A splice variants.
  • a summary graph is shown which illustrates the 100 ⁇ M PS induced increase in the response to 200 ⁇ M NMDA + 10 ⁇ M glycine for oocytes expressing NR1 011 /2A or NRl ⁇ n /2A receptors.
  • the data were normalized relative to the current response induced by co-application of 200 ⁇ M NMDA + 10 ⁇ M glycine to the same oocyte.
  • Error bars represent S.E.M of the number of oocytes indicated in parantheses .
  • FIG. 22 contains bar graphs of data indicating PS induced modulation on NRl subunits is pH dependent.
  • the data were normalized relative to the current response induced by co-application of 200 ⁇ M NMDA + 10 ⁇ M glycine to the same oocyte.
  • Error bars represent S.E.M of the number of oocytes indicated in parantheses . (Significance is indicated for NR1 011 /2A by * : p ⁇ 5xl0 "5 and for NR1 011 /2A by * : p ⁇ 5xl0 "6 and ** : p ⁇ 5xl0 "7 ) .
  • Figure 23 is a multiple sequence alignment of the ligand binding domains of the human retinoic acid receptor ⁇ (RAR) , the human retinoid X receptor a (RXR- a) , the human progesterone receptor (PR) , the human glucocorticoid receptor (GR) , the human estrogen receptor (ER) and the NMDA receptor NRl subunit (NR1 Q11 ) .
  • RAR human retinoic acid receptor ⁇
  • RXR- a the human retinoid X receptor a
  • PR human progesterone receptor
  • GR human glucocorticoid receptor
  • ER human estrogen receptor
  • NR1 Q11 NMDA receptor NRl subunit
  • conserveed identical residues are bold and underlined and similar residues are bold.
  • the triangles ( ⁇ ) under the NR1 011 sequence indicate the five mutation sites in the Penta-mutant .
  • the five mutation sites are R182A, K193
  • Figure 25 is a bar graph of data indicating 3a5 ⁇ - Pregnanolone Sulfate has similar effect on NMDA receptors consisting of wild-type NRla/NR2A and Penta-Mutant/NR2A subunits.
  • the bars represent percent decrease in maximum inward currents elicited by 300 uM NMDA and 50 urn Glycine in the presence of 100 uM 3 ⁇ 5/3-Pregnanolone Sulfate.
  • 3 ⁇ f5j8-Pregnanolone Sulfate inhibits wild-type NRla/NR2A responses by 45.49 ⁇ 5.76% (mean + SEM) and Penta- Mutant/NR2A by 42.79 ⁇ 2.29%.
  • FIG. 26 contains graphical representations of data indicating inverse modulation of NMDA receptor subtypes by PS.
  • a -D are examples of traces obtained from oocytes previously injected with NR1/NR2A, NR1/NR2B, NR1/NR2C, or NR1/NR2D mRNAs. The bar indicates the period of drug application. Interval between consecutive current traces was 45 s.
  • the receptors were activated by co-application of 10 ⁇ M glycine plus 80 ⁇ M NMDA (NR1/NR2A, A) , 25 ⁇ M NMDA (NR1/NR2B, B and NR1/NR2C, C) , or 10 ⁇ M NMDA (NR1/NR2D, D) .
  • Co-application of 100 ⁇ M PS to NR1/NR2A or NR1/NR2B receptors resulted in an increase in the agonist response
  • co-application of 100 ⁇ M PS to NR1/NR2C or NR1/NR2D resulted in a decrease in the agonist response.
  • E is dose-response curves for PS effect on NR1/NR2 receptors.
  • Responses were normalized to the control response obtained by application of 10 ⁇ M glycine plus 80 ⁇ M NMDA (NR2A) , 25 ⁇ M NMDA (NR2B, NR2C) or 10 ⁇ M NMDA (NR2D) .
  • Error bars indicate SEM.
  • F indicates the effect of holding potential on modulation of the NMDA/glycine response by PS.
  • Error bars indicate SEM.
  • Figure 27 contains four dose response curve graphs.
  • the choice of NR2 subunit determines the direction of PS modulation of the glutamate and NMDA concentration- response curves.
  • Data points are averaged normalized peak NMDA-induced current responses obtained from oocytes injected with (A) NR1/NR2A, (B) NR1/NR2B, (C) NR1/NR2C, or (D) NR1/NR2D mRNAs .
  • Concentration-response data for NMDA (squares) and for -glutamate (circles) were obtained in the presence of 10 ⁇ M glycine.
  • Figure 28 contains graphical representations of data indicating that the choice of NR2 subunit determines the direction of PS modulation of the glycine concentration- response curve.
  • Data points are averaged normalized peak current responses obtained from oocytes injected with (A) NR1/NR2A, (23) NR1/NR2B, (C) NR1/NR2C, or (D) NR1/NR2D mRNAs.
  • Concentration-response data for glycine were obtained in the presence of 10 ⁇ M L-glutamate and in the absence (open triangles) and presence (closed triangles) of 100 ⁇ M PS.
  • the data for each oocyte were normalized relative to the current response induced by co- application of 200 ⁇ M NMDA plus 10 ⁇ M glycine. Error bars represent SEM.
  • Figure 29 contains graphical representations of data indicating the choice of NR2 subunit influences 3 ⁇ 5/3S inhibition of the NMDA response.
  • a -D are examples of traces obtained from oocytes previously injected with NR1/NR2A, NR1/NR2B, NR1/NR2C, or NR1/NR2D mRNAs, respectively.
  • the jbar indicates the period of drug application. Interval between consecutive current traces was 45 s.
  • the receptors were activated by co-application of 10 ⁇ M glycine plus 80 ⁇ M NMDA (NR1/NR2A, A) , 25 ⁇ M NMDA (NR1/NR2B, B and NR1/NR2C, C) , or 10 ⁇ M NMDA (NR1/NR2D, D) .
  • G is a concentration response curve showing the effect of 3a5 ⁇ S on glutamate.
  • the data for each oocyte were normalized to standard current responses induced by co- application of 200 ⁇ M NMDA and 10 ⁇ M glycine.
  • Concentration response data for glutamate alone is the same as in Fig. 27, and is repeated for comparison. H indicates the effect of 3o;5/3S on glycine concentration- response curve.
  • Data points are averaged normalized peak current responses to glycine from oocytes injected with NR1/NR2A subunits obtained in the presence of 10 ⁇ M glutamate and in the absence (s) or presence ( ⁇ ) of 100 ⁇ M 3o;5/3s.
  • the data for each oocyte were normalized to standard current responses induced by co-application of 200 ⁇ M NMDA and 10 ⁇ M glycine. Concentration response data for glycine alone is the same as in Fig. 28, and is repeated for comparison.
  • PS Pregnenolone Sulfate
  • Figure 31 contains schematic representations of wild-type NMDAR NR2B, NR2D subunits, and the NR2 chimera constructs. Fragments from NR2B are in black and fragments from NR2D are in grey. Vertical bars correspond to the three transmembrane domains and the re- entrance domain. The numbers are the positions of the ligation junctions.
  • Figure 32 is a bar graph of data indicating 100 ⁇ M Pregnenolone Sulfate modulates the inward currents elicited by 300 ⁇ M NMDA and 50 ⁇ M Glycine from Xenopus oocytes expressing recombinant heteromeric NR1 011 /NR2 NMDA receptors.
  • PS potentiates NRl 011 /NR2Bwt by 78.36 ⁇ 7.89% (mean ⁇ SEM) , NR1 Q11 /NR2B-D Chimera(I) by 52.69 ⁇ 3.58%, NR1011/NR2B-D chimera (IV) by 66.05 ⁇ 2.76%; inhibits NRl 011 /NR2Dwt by 45.18 ⁇ 2.62%, NR1 Q11 /NR2B-D Chimera (II) by 43.04 ⁇ 5.44%, and NR1 011 /NR2B-D Chimera (III) by 36.74 ⁇ 6.54%.
  • the error bar shows standard error.
  • aspects of the present invention are based on the identification of steroid molecules which exhibit selectivity with respect to NMDA receptor subunit composition. This observation indicates that it is possible to develop or identify drugs (steroid based as well as non-steroid based) that selectively target specific NMDA receptor subtypes to modulate neurotransmitter recognition site ligands.
  • One aspect of the present invention is a method for identifying a subunit specific modulator of the NMDA receptor. This is accomplished through use of a plurality of NMDA receptors which differ in their subunit identity (NRl and NR2 subunits) .
  • the plurality of NMDA receptors is then contacted with a neurotransmitter site agonist (e.g., L- glutamate or NMDA) in the presence and absence of a candidate modulator.
  • a neurotransmitter site agonist e.g., L- glutamate or NMDA
  • Each NMDA receptor is then assayed for receptor activity produced by the neurotransmitter site agonist, in the presence and absence of the candidate modulator.
  • An increase or decrease in activity of the receptor in the presence of the candidate modulator, as compared to the respective activity in the absence of the candidate modulator, indicates modulatory activity on the receptor.
  • Such an increase or decrease in activity in at least one, but not all members of the plurality of NMDA receptors is an indication that the candidate modulator is a subunit specific modulator.
  • the subunit specificity of the candidate modulator is identified by comparing the subunit identity of the subset of NMDA receptors which are subject to modulation by the candidate modulator.
  • the candidate modulator is specific for the subunits, or domains of the subunits, which are common to this subset of receptors and absent or non- functional in the remaining receptors of the plurality.
  • Subunit specificity may be for one or more particular isoforms, chimeras or mutants of either the NRl or NR2 subunit, that is to say it may be independent of one of the subunits.
  • the modulator may be specific for a particular combination of the two subunits .
  • a modulator of the NMDA receptor is defined herein as binding at a modulatory site, and may be either a positive modulator or a negative modulator, or a null modulator.
  • a positive modulator enhances agonist activity. It has been shown that positive and negative modulators of the NMDA receptor act through distinct sites. Pregnenolone sulfate is a known positive modulator of the NMDA receptor. This is often referred to in the art as a modulatory site agonist.
  • a negative modulator inhibits agonist activity. Examples of known negative modulators of the NMDA receptor are 17/3- estradiol and pregnanolone hemisuccinate (3o;5/3HS) , also referred to in the art as inverse agonists.
  • Negative modulators have been shown to be neuroprotective from otherwise lethal exposure to NMDA, whereas positive modulators enhance excitotoxicity .
  • a null modulator has no intrinsic activity in and of itself other than to inhibit the action of a positive and/or negative modulator by competitively binding at the modulatory site .
  • a neurotransmitter recognition site ligand as the term is used herein refers to a ligand (agonist or antagonist) which binds at the neurotransmitter recognition site.
  • agonists are NMDA, L- glutamate and glycine.
  • An increase or decrease in receptor activity in the presence of the candidate modulator, as compared to activity in the absence of the candidate modulator, is identified by a reproducible, statistically significant change in receptor activity in response to ligand (agonist or antagonist) .
  • Current methodology is sensitive to the detection of approximately a 5% or greater difference in receptor activity, with an 8% difference or greater being a strong indication of modulatory activity.
  • Preferably, a 10% or greater difference in receptor activity in the presence of candidate modulator is used to identify modulator activity.
  • Subunit identity refers to the specific subunit components NRl and NR2 used to generate the NMDA receptor. This term encompasses both natural and recombinantly produced subunits and receptors. Preferably, the NMDA receptors used in the method are produced recombinantly as this allows for use of a greater diversity of receptor subunit identities. There are eight known natural isoforms of the NRl subunit
  • NR1 000 , NR1 001 , NR1 010 , NR1 Q11 , NR1 100 , NR1 101 , NR1 110 , and NRl n ⁇ NR2 subunit
  • NR2A, NR2B, NR2C, and NR2D four known natural isoforms of the NR2 subunit
  • Additional isoforms of each subunit will likely be identified in future research, and their use is also encompassed by the present invention.
  • mutant isoforms e.g., point mutant, deletion mutant, etc.
  • chimeric isoforms are suitable for use in the method.
  • Mutation or other sequence manipulation of an isoform for use in the method should not result in loss of detectable function/activity of the NMDA receptor which is produced.
  • the receptor subunits used in the experiments detailed below in the Exemplification section were of human origin.
  • the present invention is not however limited to the use of human receptors. Homologous subunits or subunit regions of homologous receptors from other species are also suitable for use in the present invention.
  • NRl subunits which contain an a exon encoded protein domain. These sites correspond to residues 182, 193, 202, 233, and 252 of human NR1 Q11 .
  • residues 182, 193, 202, 233, and 252 of human NR1 Q11 may be mutated in the NRl subunit to produce the NMDA receptor for use in the present invention.
  • Preferred point mutations are R182A, K193A, K202A, R233A, and R252A.
  • the nature of the specific substituted amino acid may have an effect on mutant function. As such, substitution of any positively charged amino acid into these positions is expected to produce a mutant having a phenotype similar to the penta- mutant characterized in Example 5.
  • Chimeric NRl or NR2 subunits used in the present invention preferably retain intact protein domains.
  • Such protein domains are usually contained within individual exons, often being delimited by those exons.
  • the a exon of the NRl subunit has been identified as encoding a protein domain which is involved in subunit specific modulation of the receptor. It is likely that domains encoded by other exons will also be useful in identifying additional subunit specific modulators which recognize and/or function through those specific domains.
  • Prime candidates are the other two alternatively spliced exons, ⁇ and ⁇ , of the NRl subunit.
  • Some domains of the NR2 subunits identified as useful in the present invention correspond to residues 534-870 of human NR2B, and also residues 548-892 of human NR2D.
  • the domain which corresponds to residues 703-870 of human NR2B has been identified as necessary and sufficient for stimulation of the receptor by neurosteroids .
  • This domain confers neurosteroid stimulation to the NR2B/NR2D chimera, whereas wild type NR2D is inhibited by the same neurosteroids.
  • the identification of these domains represent the beginnings of a growing understanding of the domains involved in neurosteroid modulation.
  • the use of chimeras made of different combinations of subunit domains in the present invention will identify subunit specific modulators which function through as yet unidentified functional protein domains of the respective subunits.
  • NR2 subunits used in the method dictates the types of subunit specific modulator which is identified. For instance, it may be useful to vary the identify of one subunit, either NRl or NR2 , in combination with a fixed identity of the other subunit. Any particular combination of the NRl and NR2 subunits described herein is appropriate for use in the present invention.
  • Assaying for receptor activity can be by any method known in the art. Preferred assay systems allow expression and utilization of recombinant receptors. A preferred assay system is an oocyte expression system.
  • Candidate modulators may be obtained from a variety of sources. They may be naturally occurring molecules, or alternatively synthetic molecules. Although the modulators provided as examples in the Exemplification section below are steroid based molecules, a subunit specific modulator may also be a non-steroid based molecule.
  • Candidate modulators may be known neuromodulators, or may be otherwise derived from known neuromodulators .
  • candidate modulators are obtained from a library of small molecules, either natural or synthetic (e.g., produced by rational drug design or randomized combination) .
  • NMDA receptor subunits are differentially distributed throughout the nervous system
  • therapeutic effects can be achieved through subunit specific intervention, accomplished by targeting a specific receptor, made up of specific subunits, with a subunit specific modulator identified by the above described method.
  • targeting has use in therapeutic intervention in neurological disorders which are linked to glutamate receptor function.
  • receptors consisting of specific subunits can be targeted to inhibit the neurodegeneration which results from cerebral ischemia.
  • Specific inhibition of AMPA/kainate receptor function can also be beneficial in the treatment of ischemic stroke.
  • the neuroactive steroid class of glutamate receptor modulatory agents may also prove to be neuroprotective in brain ischemia.
  • PS Pregnenolone sulfate
  • NMDA NR1 100 + NR2A receptors electrophysiologically with respect to efficacy and potency of modulation.
  • PS is shown to reduce the efficacy of kainate without affecting its potency, when given to Xenopus oocytes expressing GluRl, GluR3 or GluR6 receptors. This indicates a noncompetitive mechanism of action on these specific receptors.
  • PS is shown to enhance the efficacy of NMDA without affecting its potency when given to oocytes expressing NR1 100 + NR2A subunits.
  • the modulatory efficacy, but not the potency, of PS is increased two- fold by co- injection of NR1 100 + NR2A cRNAs as compared with NR1 100 cRNA alone.
  • NR2A subunit controls the efficacy of neurosteroid enhancement, but not inhibition, which is consistent with previous findings that potentiating and inhibitory steroids act at distinct sites on the NMDA receptor.
  • Results presented in Example 1 of the Exemplification section below indicate that selective modulators can be developed which specifically target particular AMPA or kainate receptor subtypes. Only negative steroid modulation of recombinant AMPA and kainate receptors was observed in the experiments described in Example 1 below.
  • pregnenolone sulfate reduces the maximum kainate-induced currents without altering the kainate EC50, consistent with a noncompetitive mechanism of inhibition.
  • 100 ⁇ M pregnenolone sulfate produces over 70% inhibition of the response to kainate.
  • the same concentration of pregnenolone sulfate inhibited GluR6 receptor response to kainate by only 42%.
  • the concentration response curve for inhibition of GluR6 receptors by pregnenolone sulfate is more shallow than for GluRl or GluR3 receptors, which indicates that pregnenolone sulfate inhibits kainate and AMPA receptors by different mechanisms.
  • Such specific modulators are useful in determining the receptor composition of specific cell populations, as well as probing the functional roles of AMPA and kainate receptors in si tu (Wisden and Seeburg, J " . Neurosci . 13 : 3582-3598 (1993)).
  • the molecule of pregnenolone sulfate has hydrophobic part that could be incorporated into the membrane, while charged sulfate part remains in aqueous surrounding. According to this hypothesis the molecule of NRl protein folded so that the N-terminal insert is positioned close to the membrane surface .
  • the results detailed in Example 6 are the first to indicate that the modulatory effect of PS is contingent upon the NR2 subunit composition of the NMDA receptor, and that PS inhibits, rather than enhances, the function of NR1/NR2C and NR1/NR2D receptors.
  • a strongly electronegative center attached to C3 is therefore sufficient to support some degree of steroid inhibition of the NMDA receptor.
  • the lack of significant neuroprotection or inhibition of Ca + accumulation with 3 ⁇ ;5/3F is thought to reflect either the difficulty of detecting the effect of such a weak inhibitor, or a difference in the interaction of 3o;5/3F with native NMDA receptors of hippocampal neurons, as compared to the NR1 100 :NR2A subtype used for the electrophysiological studies.
  • R 2 is -H, -CH 3 , or is absent when ring A has three double bonds
  • R 1 is an aliphatic or aromatic group
  • R 4 is either hemioxylate, hemisuccinate or hemiglutarate.
  • the compound is either pregnanolone hemioxylate, pregnanolone hemisuccinate, or pregnanolone hemiglutarate .
  • the ability of compounds with the above structural formula to inhibit agonist activation of the NMDA receptor can be therapeutically applied to inhibit NMDA receptor mediated ion-channel activity in an individual in need thereof by administering an effective amount of the compound to the individual. Effective amounts range from about 1 to about 500 ⁇ M, with a preferred range being from about 50 to about 250 ⁇ M. Such administration can be used to inhibit the toxic effects associated with activation of the NMDA receptor in neurons of the individual.
  • Another useful application of this method of inhibiting agonist activation of the NMDA receptor is in reducing neuronal cell death resulting from L-glutamate activation of the NMDA receptor.
  • This also is achieved by administration of the compound to an individual as described above. Additional diseases which can be treated by said administration of the compound are neuropathic pain, drug withdrawal/dependency, epilepsy, glaucoma, chronic neurodegenerative diseases, amyotrophic lateral sclerosis, anxiety disorders, brain cell death, ischaemia, stroke, and trauma in an individual when said disease results from NMDA induced NMDA receptor activation.
  • Administration of the compound can also be used to inhibit the excitatory or L-glutamate-mediated synaptic activity in an individual in need thereof.
  • Another aspect of the present invention relates to a method for rationally designing steroid-based therapeutics for the treatment of stroke and disorders arising from the overactivation of the NMDA receptor.
  • the finding that a variety of negatively-charged carboxylic acid esters can substitute for the sulfate ester at the C3 position of the above described molecule offers prospects for modifying the steroid nucleus to optimize pharmacological and pharmacokinetic properties.
  • Carboxylic acid derivatives of neuroactive steroids should offer improved penetration into the CNS and reduced susceptibility to hydrolysis by sulfatases.
  • PS inhibits AMPA- and kainate-induced currents recorded from chick spinal cord neurons (Wu et al . , Mol . Pharmacol . 40 : 333-336 (1991)).
  • onset of inhibition is rapid ( ⁇ 10 s) , strongly suggests a direct interaction between PS and the AMPA/kainate receptor.
  • PS 100 ⁇ M reduced the kainate I ma ⁇ by 83%, while the kainate EC 50 was unchanged (18 ⁇ M vs. 23 ⁇ M with PS) .
  • GluR3 AMPA receptor function was also markedly reduced by PS, which reduced the kainate I ma ⁇ by 71% without altering the EC 50 (27 ⁇ M vs. 32 ⁇ M in the presence of PS) .
  • PS also appeared to act as a noncompetitive inhibitor of GluR6 kainate receptor function, reducing the kainate J ma ⁇ by 33% with no change in EC 50 (545 nM vs. 567 nM in the absence and presence of PS, respectively) .
  • PS Inhibition by PS was dose-dependent, with EC 50 's of 43 ⁇ M and 12 ⁇ M at the GluRl and GluR3 receptors, respectively (Fig. 2F) .
  • the extent of inhibition of GluR6 kainate receptors was considerably less than for the GluRl or GluR3 AMPA receptors.
  • Table 1 tabulates the effects of a series of steroids on kainate responses mediated by GluRl, GluR3 , or GluR6 receptors.
  • the GluR6 kainate receptor tended to be less sensitive than GluRl and GluR3 AMPA receptors to inhibition by steroids.
  • Holding potential during drug application is -1UU mV All steroids were applied at 100 ⁇ M Oocytes injected with the indicated subunit cRNA were pre-equilibrated with steroid for 10 s prior to co-application of steroid and NMDA or kainate Values are mean percentage change ⁇ S E in the response to 100 ⁇ M NMDA (NR1 I00 + NR2A) or 100 ⁇ M (GluRl and GluR3) or 10 ⁇ M (GluR6) kainate
  • the number of experiments is given in parentheses * Indicates a statistically (P ⁇ 0 05) change in the induced current
  • Steroid modulation was rapid in onset, and the degree of modulation was similar whether NMDA was applied simultaneously with the steroid modulator or after a 10 s preincubation with steroid. Reversal of steroid modulation was also rapid, with NMDA responses returning almost to control levels after a 30 s wash (Fig. 3A and B) .
  • the potentiating effect of PS was due primarily to an increase in NMDA efficacy, which was nearly tripled in the presence of 100 ⁇ M PS, with no change in the NMDA EC 5Q .
  • the EC 50 for enhancement of the 100 ⁇ M NMDA response by PS was 32 ⁇ M (Fig. 3D) , with a maximum enhancement of 343%.
  • Pregnenolone hemisuccinate also potentiates the NMDA response (Table 1) , whereas 3/35/3S had an inhibitory effect similar to 3 ⁇ 5/3s, although it was slightly less potent (Fig. 3D) .
  • oocytes injected with NR1 100 cRNA were similar to those injected with NR1 100 plus NR2A cRNA with respect to the potency and efficacy of the inhibitory steroids 3O5/3S and 3/35/3S .
  • Plasmids were obtained from the following individuals: Dr.tician Zukin (Albert Einstein College of Medicine, Bronx, NY), NR1 100 ; Dr. Shigetada Nakanishi (Kyoto University Faculty of Medicine, Kyoto, Japan) , NR2A; Dr. Stephen Heinemann (Salk Institute, San Diego, CA) , GluRl (flop), GluR3 (flop), and GluR6.
  • Plasmids were linearized with BamHI (NR1 100 ) , Xhol (NR2A, GluRl, GluR3) or Xjbal (GluR6) prior to in vi tro transcription with T3 (GluRl, GluR3 , GluR6) or T7 ( R1 100 , NR2A) RNA polymerase using the Message Machine kit (Ambion, Austin,
  • Oocytes were harvested and maintained in ND96 (NaCl, 96 mM; MgCl-. 1 mM; KC1, 2 mM; CaCl 2 , 1.8 mM; HEPES, 5mM; pyruvate, 2.5 mM; 50 mg/ml gentamicin) according to published protocols (Stuhmer and Parekh, Electrophysiological recordings from Xenopus oocytes, in: B. Sakmann, E. Neher (Eds.), Single-Channel Recording, 2nd edn., Plenum Press, New York, 1995, pp. 341-356).
  • RNA solutions (50-70 nl) were injected into oocytes for delivery of NR1 100 (5 ng) , NR1 10Q :NR2A (1:10 ng) , GluRl (5 ng) , GluR3 (5 ng) , or GluR6 (20 ng) .
  • Oocytes were incubated in ND96 at 18-20°C for 3-7 days prior to electrophysiological recording.
  • Steroids were obtained commercially from Steraloids, (Wilton, NH) and Sigma (St. Louis, MO), except 3 ⁇ ;5/3S, 335 ⁇ ;S, and 3/35/3S, which were provided by Dr. Robert Purdy (Dept . of Psychiatry, University of California, San
  • Steroid solutions were prepared as 70-100 mM frozen stocks in dimethyl sulfoxide (DMSO) .
  • DMSO dimethyl sulfoxide
  • the final DMSO concentration of all recording solutions was between 0.1% to 0.5% and was constant within an experiment. Over this concentration range, DMSO alone does not affect kainate or NMDA responses.
  • Perfusion control and data acquisition were carried out using an automated oocyte electrophysiology workstation that was developed for the rapid collection of concentration-response data and real-time waveform analysis (Yaghoubi et al . , Soc . Neurosci . Abstr. 20 : 1109 (1995)).
  • This system is implemented on a Macintosh Ilci computer (Apple Computer, Cupertino, CA) equipped with MacA-DIOS II data acquisition and control hardware (GW Instruments, Somerville, MA) .
  • Drug application is controlled by the computer through a series of valves, using custom-written software based on the SuperScope II development environment, improving the efficiency and reproducibility of data collection.
  • PS concentration-response curves for GluRl and GluR3 experiments were determined at -80 mV due to high levels of receptor expression which resulted in off-scale current responses at -100 mV. Current and voltage recordings were digitized at 100 Hz for analysis. GluR6 injected oocytes were treated for 10 min with 10 ⁇ g/ml concanavalin A to prevent fast desensitization.
  • NMDA and kainate responses by steroid compounds in expressed as percentage change, [ (T'/J) -1] X 100 %, where J is the average of four control responses, obtained from an oocyte before application and after washout of steroid, and I ' is the average of two responses obtained from the same oocyte in the presence of steroid.
  • Peak NMDA responses were normalized to the maximal (100 ⁇ M) NMDA response for analysis and comparison except where otherwise specified.
  • Peak kainate- induced currents are normalized to the maximal (100 ⁇ M for GluRl and GluR3 receptors; 10 ⁇ M for GluR6 receptors) kainate response.
  • Kainate rather than AMPA, was used to activate GluRl and GluR3 receptors, because desensitization was much less with kainate than with AMPA.
  • oocytes were pre-equilibrated with steroid modulator for 10 s prior to application of NMDA, but this preincubation was found to have little if any effect on the extent of modulation of the peak NMDA- induced current, and the preincubation period was therefore omitted in subsequent experiments. Results are expressed as mean ⁇ S.E.M. Statistical comparisons were made using Student's t-test.
  • the Ca 2+ -sensitive dye Fluo-3, AM and a trypan blue exclusion assay were employed to measure NMDA-induced Ca 2+ influx and cell death, respectively, in primary cultures of rat hippocampal neurons .
  • a number of pregnane and pregn-5-ene steroids were examined for their ability to alter the increase in [Ca 2+ ] i and the neuronal death produced by NMDA exposure .
  • 3 ⁇ 5/3S protects neurons from the cell death produced by acute (15 min) exposure to NMDA, raising the EC 50 for NMDA-induced neuronal death from 28 ⁇ M to 71 ⁇ M and lowering the maximal NMDA-induced excitotoxicity from 80% to 63% cell death (Fig. 4B) .
  • SR-95531 a selective GABA A receptor antagonist (Farrant M, and Webster RA (1989) GABA antagonists: their use and mechanisms of action, in Drugs as Tools in Neurotransmi tter Research (Boulton AA, Baker, GB and Juorio, AV, eds.) pp 161-219, Humana Press Inc., Clifton) have no effect on NMDA-induced neuronal death (not shown) , arguing that the GABA A , AMPA, and kainate receptor types do not play a significant role in this process .
  • a series of carboxylic acid derivatives of 3cx5 ⁇ were synthesized (Fig. 7A) .
  • the three negatively-charged derivatives, pregnanolone hemioxylate (3 ⁇ ;5/3HO) , pregnanolone hemisuccinate (3 ⁇ 5/3HS) , and pregnanolone hemiglutarate (3o;5/3HG) are about equally effective in inhibiting I NMDA/ the NMDA-induced current of Xenopus oocytes expressing NR1 100 + NR2A subunits, but the degree of inhibition is substantially less than is observed with 3 ⁇ 5/3S (Figs.
  • the uncharged pregnanolone formate (3 ⁇ :5/3F) significantly inhibits I NHDA , but is significantly less effective than the dicarboxylic acid derivatives, while 3o ⁇ 5 ⁇ itself is inactive. Consistent with these results, the negatively- charged 3o;5/3HO, 3 ⁇ 5/3HS, and 3 ⁇ 5j ⁇ HG inhibit the NMDA- induced rise in [Ca 2+ ] i by approximately 40% in primary hippocampal cultures, while the uncharged 3 ⁇ ;5/3 and 3o;5/3F have no significant effect on NMDA-induced Ca 2+ accumulation (Fig. 7C) .
  • PS is a potent positive modulator of NMDA receptor function (Wu et al . , Mol Pharmacol 40 : 333-336 (1991); Bowlby MR, Mol Pharmacol 43 : 813-819 (1993)).
  • PF pregnenolone formate
  • PHO pregnenolone hemioxylate
  • PHS pregnenolone hemisuccinate
  • PG pregnenolone hemiglutarate
  • PHO NMDA-induced excitotoxicity
  • 3 ⁇ 5/3S is also demonstrated to markedly inhibit NMDA-induced changes in [Ca 2+ ] and neuronal death under both acute and chronic exposure conditions, consistent with previous finding that 3o;5/3s inhibits NMDA-induced currents in neurons maintained in primary culture (Park-Chung, et al . , Mol Pharmacol 46 : 146-150 (1994)). It is interesting that, in acute treatments, 3 ⁇ 5 ⁇ S reduces both the NMDA EC 50 and E ma ⁇ for causing cell death, while only reducing the E ma ⁇ in chronic treatments. The reason for this difference is unclear, but may indicate metabolic conversion of 30(5/35, such as through the action of a steroid sulfatase, during the course of the chronic treatment, or an adaptive change at the NMDA receptor itself.
  • 3 ⁇ 5/3 and stereoisomers of 3 ⁇ 5/3S were assayed for activity, as were several related synthetic pregnane steroids.
  • 3a5 ⁇ S is as effective as 3c ⁇ 5/3S at protecting against the neuronal death produced by acute exposure to NMDA.
  • the stereochemistry at C3 is not critical for inhibition of NMDA-induced neuronal death by the C53 pregnane isomers.
  • the isomers with C5 ⁇ stereochemistry exhibit reduced neuroprotective activity as compared to the C5/3 isomers.
  • Formate esters were prepared by treating a solution of the steroid (400 mg) in dry dichloromethane (30 ml) with triethylamine (2.4 ml), 4-dimethylpyridine (160 mg) and formic acid (0.32 ml) . The mixture was cooled to -20 °C and acetic anhydride (1.9 ml) added dropwise over a 30 min period with stirring. The reaction mixture was then warmed to 0 °C for 30 min and then the reaction stopped by the addition of methanol (1.0 ml) . After evaporation of the solvents in vacuo, the residue was partitioned between ethyl acetate (10 ml) and aqueous IN HCl .
  • the organic phase was washed twice with IN HCl (10 ml) and water (10 ml), and evaporated to dryness .
  • the product was crystallized twice from a mixture of acetone and hexane .
  • the hemioxalate esters were prepared as described above, using 568 mg oxalic acid instead of formic acid.
  • the hemiglutarate esters were prepared as follows: To a solution of steroid (400 mg) in dry pyridine (6 ml) was added glutaric anhydride (400 mg) and 4-pyrolidinopyridine (40 mg) . The mixture was allowed to stand at room temperature in the dark for 4 days, when thin-layer chromatography showed complete disappearance of steroid starting material.
  • the reaction mixture was then poured into ice water (20 ml) and the product extracted with ethyl acetate (20 ml) , and the extract washed with aqueous IN HCl (5 ml) and water (5 ml) . After drying the ethyl acetate solution over anhydrous sodium sulfate, the product was treated with activated charcoal (200 mg) and crystallized from a mixture of ethyl acetate and hexane .
  • the hemisuccinate esters were prepared as described above, except that succinic anhydride (225 mg) was used in place of glutaric anhydride, and the reaction required 7 days for completion at room temperature.
  • Steroids were initially dissolved in 100% DMSO, then diluted into assay buffer at a final DMSO concentration of 0.5% and sonicated for 20 min. All other solutions also contained 0.5% DMSO. Except where otherwise noted, the final steroid concentration was 100 ⁇ M. 3 ⁇ 5/3 and pregnenolone were used at 50 ⁇ M and 20 ⁇ M, respectively, as higher concentrations tended to precipitate in the assay buffer.
  • hippocampal cells were dissociated by trituration in Ca 2+ /Mg 2+ -free Hank's basic salt solution (Gibco) supplemented with 4.2 mM bicarbonate, 1 mM sodium pyruvate, 20 mM HEPES, 3 mg/ml bovine serum albumin (BSA) . Dissociated cells were then pelleted by centrifugation (500 x g, 4 min) .
  • Ca 2+ /Mg 2+ -free Hank's basic salt solution Gibco
  • BSA bovine serum albumin
  • the resulting pellet was suspended in Dulbecco's modified eagle medium (DMEM) (Gibco) supplemented with 2.4 mg/ml BSA, 26.5 mM sodium bicarbonate, 1 mM sodium pyruvate, 20 mM HEPES, 10% fetal bovine serum (FBS) (Gibco) , 100 units/ml penicillin, 100 ⁇ g/ml streptomycin (Gibco) , and a modification of Brewer's B16 defined components (with 250 nM vitamin B12 and without catalase, glutathione, and superoxide dismutase) (Pike et al . , J Neurosci 13 : 1676-1687 (1993)) .
  • DMEM Dulbecco's modified eagle medium
  • Cells were then plated onto poly-L-lysine- coated 24 -well culture dishes (Nunclon) at a density of 15,000 cell/cm 2 and maintained in a humidified atmosphere containing 5% C0 2 /95% air at 37°C. After 48 h, nonneuronal cell division was inhibited by a 48 h exposure to 1 ⁇ M cytosine arabinoside. Cultures were subsequently maintained in serum-free DMEM plus defined components and were used for experiments 16 to 24 days after plating.
  • NMDA-induced increases in [Ca 2+ ] i were measured with the Ca 2+ - sensitive fluorescent dye Fluo-3, AM (Molecular Probes) and a Cytofluor 2350 (Perceptive Biosystems) fluorescence plate reader, using excitation and emission filters of 485 nm and 530 nm, respectively.
  • Hippocampal neurons were loaded with dye by incubating cultures with 10 ⁇ M Fluo-3, AM and 0.05% (w/v) Pluronic F-127 (Molecular Probes), a nonionic detergent, for 2 hours at 37°C. Fluo-3, AM and Pluronic F-127 were dissolved in dimethyl sulfoxide (DMSO, final concentration 0.5%).
  • DMSO dimethyl sulfoxide
  • NMDA Steroid or vehicle (0.5% DMSO) was added 10 min prior to the addition of NMDA.
  • NMDA was dissolved in DMEM; steroids and A-23187 were dissolved in DMSO.
  • DMSO was also added to controls to maintain a constant final DMSO concentration of 0.5. Data are expressed as the percentage change in the NMDA-induced increase in [Ca ⁇ ] i in the presence of the indicated steroid. None of the steroids tested significantly altered [Ca 2+ ] j _ in the absence of NMDA.
  • NMDA-induced Cell Death Primary cultures of rat hippocampal neurons were exposed to NMDA (dissolved in DMEM) for 15 min (acute exposure) or 16 hours (chronic exposure) . In acute exposure experiments, cultures were treated with steroid, MK-801 (Research Biochemicals International; dissolved in DMEM), or vehicle during and/or after NMDA exposure. Steroids were dissolved in DMSO (0.5% final concentration), and all treatment media contained 0.5% DMSO.
  • Percent cell death is expressed as follows: (number of trypan blue stained neurons) / (total number of neurons) x 100%.
  • the basal level of neuronal death (termed background) was 0-10% 4 and was subtracted from the raw data in each experiment .
  • NR1 100 (NR1G) and NR2A clones were kindly provided by Dr. R. S. Zukin (Albert Einstein College of Medicine, New York, NY) and Dr. S. Nakanishi (Faculty of Medicine, Kyoto University, Kyoto, Japan), respectively.
  • mRNA was prepared through in vi tro transcription of NR1 100 and NR2A cDNAs using the mMessage mMachine kit (Ambion, TX) . On the day following isolation, batches of 20-40 selected Xenopus oocytes were injected with 50 nl of prepared RNA solution (0.5 ng NR1 100 and 5 ng of NR2A mRNA/oocyte) . Oocytes were maintained in glass petri dishes containing Barth's solution (84 mM NaCl, 1 mM KCl, 2.4 mM NaHC0 3 , 0.82 mM
  • Example 3 Figure 11 shows a comparison of traces of NMDA induced responses obtained from oocytes expressing NR1 100 /NR2A receptors bathed in normal (Ca 2+ containing) Ringer solution and Ba-Ringer solution in which Ca 2+ was replaced with Ba 2+ .
  • Current traces obtained in Ba-Ringer solution do not exhibit the rapidly inactivating component that is seen in normal Ringer, and which most likely reflects current through Ca 2+ activated CI " channels (Leonard et al . , Neuron 4 : 53-60 (1990)). All further experiments were performed in Ba-Ringer.
  • L-glutamate, glycine and NMDA EC 5Q s for NRl isoforms lacking N-terminal insert were less than ones for isoforms with N-terminal insert (except for NR1 001 , see Table 2) .
  • the absolute current responses induced by saturating concentration of agonists 500 mM NMDA and 10 mM glycine) were in the range from 800 to 1800 nA.
  • oocytes that showed current responses in the range of 0.1 ⁇ Amp to 1 ⁇ Amp induced by 50 mM (or 80 mM NMDA for isoforms with N-terminal insert) and 10 mM glycine were used to assess the effects of pregnenolone sulfate and pregnanolone sulfate on NMDA receptors comprised of different isoforms of NRl subunit.
  • NR1 011 /NR2A 0.56 ⁇ 0.03 (0.39 ⁇ 0.03) 1.17 ⁇ 0.03 (1.13 ⁇ 0.08) 1.59 ⁇ 0.03 (2.08 ⁇ 0.21)
  • R1 111 / R2A 0.71 ⁇ 0.06 (0.51 ⁇ 0.03) 1.39 ⁇ 0.02 (1.58 ⁇ 0.09) 1.87 ⁇ 0.12 (2.62 ⁇ 0.27)
  • NRl n ⁇ /NR2B 24 ⁇ 1 1.52 ⁇ 0.10 1.20+0.10
  • NRl isoforms were similar, revealing PS EC 50 s that ranged from 25 to 45 mM.
  • the maximum extent of inhibition obtained for different NRl isoforms ranged from 75 ⁇ 7% (for NRl ⁇ /NR2A) to 99 ⁇ 3% (for NR1 100 /NR2A, see Table 4) .
  • the differences in maximum inhibition were insignificant and no apparent correlation with the presence of N-terminal insert was observed.
  • Table 5 shows alternatively spliced exons present in each NRl splice variant.
  • PS pregnenolone sulfate
  • NMDA dose response of NR1 001 /NR2A and NR1 111 /NR2A splice variants were determined, in the presence or absence of 100 ⁇ M PS ( Figure 15) .
  • the Emax obtained for NR1 011 /NR2A in the presence of absence of PS was 1.79 and 1.2 respectively, while for NRl n ⁇ /NR2A was 2.7 and 1.33.
  • NMDA dose response curves of NR1 011 /NR2A and NR1 111 /NR2A splice variants were constructed at pH 8.5 , in the presence or absence of 100 ⁇ M PS ( Figure 19) .
  • the extrapolated maxima (Emax) obtained from the concentration-response, indicate that the relative current change of each splice variant is pH dependent.
  • South African clawed frogs Xenopus laevis
  • Xenopus laevis were purchased from either Xenopus One (Ann Arbor, MI) or Nasco (Fort Atkinson, WI) .
  • Frog brittle is also purchased from the same vendors .
  • Plasmids containing the NR1 100 , NR1 Q00 , NRl ⁇ , NR1 Q11 , NR1 101 , NR1 Q01 inserts (encoding different NMDA receptor splice variants) and NR2A (encoding NMDA receptor subunit) were kindly provided by Dr. Shigetada Nakanishi of Kyoto University Faculty of Medicine, Kyoto, Japan.
  • Competent DH5 ⁇ cells (used for transformation) were purchased from Gibco (Gaithersburg, MD) . Maxiprep columns were purchased from Qiagen (Chatsworth, CA) . Restriction enzymes were purchased from New England Biolabs (Beverly, MA) and Pharmacia (Piscataway, NJ) .
  • mMessage mMachine in vitro transcription kit (T7 RNA polymerase) was purchased from Ambion (Austin, TX) .
  • NMDA and glycine were purchased from Sigma Chemical Co. (St. Louis, MO). Chemicals used for the preparation of ND96 and Ringer solutions were purchased from Sigma and Gibco. PS and 3 ⁇ 5/3S were purchased commercially from Steraloids (Wilton, NH) and Sigma.
  • Data acquisition and instrument control hardware was purchased from GW Instruments (Somerville, MA) .
  • SuperScope II vl.43 software was also purchased from GW instruments.
  • Miniature teflon-coated valves were purchased from LeeValves (Essx, CT) .
  • Apple Macintosh Ilic was purchased from Apple Computer Inc. (Cupertino, CA) .
  • a microprocessor-controlled fixed or variable volume microinjector was purchased from Drummond Inc. (Broomall, PA) .
  • Micromanipulators (no. M3301) were purchased from World Precision Instruments (Sarasota, FL) , translation stages (single axis, no. 423) , 50mm open loop motorized DC actuators (no. 860-A) , hand-held single axis controller (no. 861) , low profile magnetic base (no. 150), 45° angle bracket (no. 360-45) from Newport Inc. (Irwine, CA) and the headstages for the electrodes from Drummond Inc.
  • the OC-725 Oocyte Clamp amplifier was purchased from Warner Instrument Corp.
  • a programmable horizontal Flaming- Brown micropipette puller was purchased from Sutter Instruments Co., CA.
  • Perfusion manifolds no. ML-6
  • PE polyethylene tubing
  • Glass pipettes for RNA microinjection RNase-free 3.5-in. glass capillaries no. 3-000-203 -Cdx
  • Glass electrodes borosilicate glass 100 ⁇ l disposable micropipettes, no. TWI50F-4) were purchased from World Precision Instruments.
  • mRNA was prepared from lyophilized plasmid pellets containing cDNA insets encoding the specific NMDA receptor splice variant. Transformation of competent DH5a (sub-cloning quality) cells resulted in bacterial cultures expressing the desired plasmids. Overnight cultures (500 ml) of transformed bacterial cells were prepared in order to grow sufficient quantities for maxiprep isolation of the plasmid cDNA.
  • plasmid cDNA was treated with proteinase K (degradation of RNAse) and extracted with a combination of phenol/chloroform/isoamyl alcohol, after which ethanol precipitation was used to pellet DNA prior to the start of the transcription reaction.
  • cDNA quality was assessed by agarose-gel electrophoresis (1.5% agarose gel). The samples were stored at - 20°C until use.
  • Electrophysiological recordings of ion currents from oocytes expressing the desired NMDA receptor subtypes were carried out using either the OC-725 Oocyte Clamp amplifier from Warner Instruments or the Axoclamp-2A amplifier from Axon Instruments Inc. in a two-electrode voltage clamp mode.
  • Intracellular recording microelectrodes were fabricated from 100 ⁇ l borosilicate glass disposable micropipettes using a 2 -step protocol on a standard horizontal Flaming-Brown micropipette puller.
  • Perfusion control and data acquisition were carried out using an automated oocyte electrophysiology workstation and custom-written software implemented in the SuperScope II environment, which was developed for the rapid collection of concentration-response data and real time waveform analysis (Yaghoubi et al . , Evaluation on neurosteroid modulation of kainate receptors using an automated system for oocyte electrophysiology . In Society for Neuroscience, pp. 1109 (1995)). All experiments were performed at room temperature (22-24°C) .
  • Sulfated steroids can act as positive or negative modulators of NMDA receptor function.
  • pregnenolone sulfate PS
  • an abundant neurosteroid potentiates NMDA-induced currents
  • 3 ⁇ 5/3- pregnanolone sulfate 3o;5/3S
  • PS does not competitively inhibit the binding of 3 ⁇ 5/3s to the NMDA receptor, indicating the presence of independent binding sites or pathways for negative and positive modulation.
  • the sequence of the NRla subunit was aligned with that corresponding to the ligand binding domain of five nuclear receptors: human retinoid X receptor a (RXR) , the human retinoic acid receptor ⁇ (RAR) , the human progesterone receptor (PR) , the human glucocorticoid receptor (GCR) and the estrogen receptor (ER) .
  • RXR human retinoid X receptor a
  • RAR human retinoic acid receptor ⁇
  • PR human progesterone receptor
  • GCR human glucocorticoid receptor
  • ER estrogen receptor
  • the mutant Penta-mutant was constructed by replacing five positively charged residues (R187, K193, K202, R233, and R252) with alanines (Fig. 23) .
  • PM was co-expressed with NR2A in the xenopus oocyte expression system.
  • the receptors containing PM were functional with an EC50 of 55.0 ⁇ 3.4 uM for the NMDA response, similar to the wild- type receptor.
  • the PM containing receptors showed a 40% reduction in sensitivity to PS in the presence of lOOuM PS, 300uM NMDA and 50 uM Glycine (Fig. 24) .
  • Mutagenesis Site directed mutagenesis was generated by a method involving PCR.
  • oligonucleotides were synthesized containing the mutation, and these oligonucleotides were used in combination with other oligonucleotides in PCR amplifications of fragments of the cDNA.
  • the product of the PCR reactions were cut with two different restriction enzymes to generate a cassette containing the mutation.
  • This cassette was then ligated into the cDNA that was cut with the same two restriction enzymes.
  • single isolates were selected, and the entire region of the amplified cassette was sequenced to check for the mutation and to insure against second- site mutations.
  • RNA Preparation of RNA. Plasmids containing the NR1 Q11 and NR2A cDNA inserts were kindly provided by Dr. Nakanishi (Kyoto University Faculty of Medicine, Kyoto, Japan) , while plasmids containing the NR2B, NR2C and NR2D cDNA inserts were kindly provided by Dr. Seeburg (Heidelberg University, Heidelberg, Germany) . Plasmids were linearized with appropriate restriction enzyme prior to in vi tro transcription using the Message Machine kit (Ambion, Inc., Austin, TX) . Expression in Xenopus oocytes .
  • oocyte positive Xenopus laevis frogs were purchased from Xenopus I (Dexter, MI) . Following 45 min of 0.15% Tricaine anesthesia, ovarian sections containing the follicular oocytes were removed from the frog through a lateral abdominal incision and were immediately placed in a calcium-free solution (NaCl 96 mM, MgCl 2 1 mM, KCl 2 mM, Hepes 50 mM, pyruvate 2.5 mM, 0.1 mg/ml gentamicin, pH 7.4) . Following 1.5 - 2 hours incubation in 0.2% collagenase (type II, Sigma Chemical Co., St.
  • the transcripts were injected at a ratio of 0.125/1.25 ng mRNA per oocyte for NRla/NR2A or penta-mutant/NR2A receptors and 0.5/5 ng mRNA per oocyte for NRla/NR2B, NRla/NR2C, and NRla/NR2D receptors.
  • the injected oocytes were used for experiments following 1-5 days of incubation in Barth's solution at 18-20°C.
  • Electrophysiology Measurements of ion currents from oocytes expressing NMDA receptors were performed using an Axoclamp-2A voltage clamp amplifier (Axon Instruments, Inc., Foster City, CA) in two-electrode voltage clamp mode.
  • the microelectrodes were fabricated with a programmed puller (Sutter Instrument Co., CA) from borosilicate glass capillaries and were filled with 3 M KCl solution. The resistance of filled microelectrodes was in the range of 1-3 M ⁇ .
  • the oocyte recording chamber was continuously perfused with Mg 2+ -free Ba-Ringer solution (NaCl 96 mM, KCl 2 mM, BaCl 2 1.8 mM, Hepes 5 mM) .
  • Mg 2+ -free Ba-Ringer solution NaCl 96 mM, KCl 2 mM, BaCl 2 1.8 mM, Hepes 5 mM
  • the perfusion solutions used did not contain Ca 2+ ions. If not mentioned otherwise, during data acquisition oocytes were clamped at a holding potential of -70 mV.
  • the membrane current was filtered at 500 Hz and sampled at 100 Hz. Drugs were applied using a gravity driven external perfusion system.
  • the working volume of the recording chamber was 30 ⁇ l and the rate of the perfusion was 50 ⁇ l/sec.
  • the drug application lasted 10 sec and was followed by a 60 sec wash.
  • the data acquisition and external perfusion control were done using the SuperScopell software package (GW Instruments, MA) . All experiments were performed at room temperature (22-24°C). Peak or steady-state current measurements were normalized and expressed as a fraction of the peak or steady-state control current measurements, which were performed before and after application of every single consentration of agonist or steroid.
  • Steroids were obtained from Steraloids, Inc. (Wilton, NH) and were dissolved in DMSO prior to use. Other compounds were obtained from Sigma (St. Louis, MO) .
  • NMDA receptor expression in Xenopus oocytes To investigate the influence of NMDA receptor subunit composition on the modulatory effects of neuroactive steroids, mRNA coding for the NR1 100 subunit was coinjected into Xenopus laevis oocytes along with mRNA coding for either the NR2A, NR2B, NR2C, or NR2D subunit. All 4 subunit combinations resulted in expression of functional NMDA receptors 1-5 days after injection, as indicated by an inward current in response to application of 80 ⁇ M NMDA plus 10 ⁇ M glycine. Concentration-response studies showed that the NMDA EC 50 differed substantially across the different subunit combinations.
  • PS was applied to oocytes expressing NR1/NR2A, NR1/NR2B, NR1/NR2C, or
  • NR1/NR2D NR1/NR2D, together with 10 ⁇ M glycine and a concentration of NMDA approximately equivalent to its EC 50 for that subunit combination.
  • the glutamate, NMDA, and glycine concentration-response curves were determined in the presence and absence of PS. As shown in Figs. 27 and 28, the nature of the modulatory effect of PS depended not only upon subunit combination, but also upon the specific agonist used. With NR1/NR2A receptors, PS enhanced the efficacy of NMDA, glutamate (Fig. 27A) and glycine (Fig. 28A) , while decreasing the glutamate and glycine EC 50 s (Table 4A) .
  • PS enhanced both potency and efficacy of NMDA, but only enhanced the potency of glutamate (Fig. 27B) , while glycine potency was increased with only a slight increase in efficacy (Fig. 28B and Table 4B) .
  • Inhibitory potency of 3 ⁇ 5/3S depends upon the NR2 subunit As shown in Fig. 29A-D, 100 ⁇ M 3o;5/3S reversibly inhibited NMDA-induced currents of Xenopus oocytes expressing NR1/NR2A (Fig. 28A) , NR1/NR2B (Fig. 28B) , NR1/NR2C (Fig. 28C) , or MR1/NR2D (Fig. 28D) receptors. However, the extent of inhibition was significantly greater with NR1/NR2C and NR1/NR2D receptors than with NR1/NR2A and NR1/NR2B receptors. Concentration-response analysis (Fig. 29A-D)
  • concentration-response curves were constructed for glutamate (in the presence of 10 ⁇ M glycine) and glycine (in the presence 10 ⁇ M glutamate) in the presence and absence of 100 ⁇ M 3 ⁇ 5/3S.
  • 3 ⁇ 5/3S decreased the efficacy with which glutamate and glycine activate NR1/NR2A (Fig. 29G-H) , NR1/NR2B (Fig. 29I-J) , NR1/NR2C (Fig. 29K-L) , and NRl/NR2D (Fig. 29M-N) receptors .
  • RNA Preparation of RNA. Plasmids containing the NR1 100 (NR1G) and NR2A cDNA inserts were kindly provided by Dr. Nakanishi (Kyoto University Faculty of Medicine, Kyoto, Japan) . Plasmids containing the NR2B, NR2C and NR2D cDNA inserts were kindly provided by Dr. P. Seeburg
  • Plasmids were linearized with appropriate restriction enzyme prior to in vi tro transcription using the Message Machine kit (Ambion, Inc., Austin, TX) .
  • NMDA receptor expression in Xenopus oocytes Female, oocyte-positive Xenopus laevis frogs were purchased from Xenopus I (Dexter, MI). Following 45 min of 0.15% Tricaine anesthesia, ovarian sections containing the follicular oocytes were removed from the frog through a lateral abdominal incision and were immediately placed in a calcium-free solution (NaCl 96 mM, MgCl 2 1 mM, KCl 2 mM, Hepes 50 mM, pyruvate 2.5 mM, 0.1 mg/ml gentamycin, pH 7.4) .
  • Oocytes were injected with 50 nL of RNA solutions using an electronic microinjector (Drummond Inc., Broomall, PA).
  • the transcripts were injected at a ratio of 0.125/1.25 ng mRNA per oocyte for NR1/NR2A receptors and 0.5/5 ng mRNA per oocyte for NR1/NR2B, NR1/NR2C, and NR1/NR2D receptors.
  • the injected oocytes were used for experiments following 1-5 days of incubation in Barth's solution at 18-20°C.
  • Electrophysiolog Measurements of ion currents from oocytes expressing NMDA receptors were performed using an Axoclamp-2A voltage clamp amplifier (Axon Instruments, Inc., Foster City, CA) in two-electrode voltage clamp mode.
  • the microelectrodes were fabricated from borosilicate glass capillaries with a programmed puller (Sutter Instrument Co., CA) .
  • Microelectrode resistance was 1-3 M ⁇ _when filled with 3 M KCl.
  • the oocyte recording chamber was continuously perfused with Mg 2+ -free Ba-Ringer solution (NaCl 96 mM, KCl 2 mM, BaCl 2 1.8 mM, Hepes 5 mM) .
  • Ba-Ringer was used to prevent NMDA receptor currents from being complicated by activation of Ca 2+ dependent CI " channels (Leonard and Kelso, Neuron . 4 : 53- 60 (1990) ) . Potentiation of the NMDA-induced current of NR1 100 /NR2A receptors by PS in Ba-Ringer tended to be less than previously observed with using Ca 2+ -containing solution (Yaghoubi et al . , Brain Res . 803 : 153-160
  • oocytes were clamped at a holding potential of -70 mV during data acquisition.
  • the membrane current was filtered at 500 Hz and sampled at 100 Hz.
  • Drugs were applied using a gravity-driven external perfusion system.
  • the working volume of the recording chamber was 30 ⁇ l and the rate of the perfusion was 50 ⁇ l/sec.
  • the drug application lasted 10 sec and was followed by 60 s wash.
  • Data acquisition and external perfusion were controlled using custom-written software implemented in the SuperScope II development environment (GW Instruments, MA) . All experiments were performed at room temperature of 22-24°C. Peak or steady-state current measurements were normalized and expressed as a fraction of the peak or steady-state control current measurements, which were performed before and after application of every single concentration of agonists or steroids .
  • PS- induced changes in E ⁇ a ⁇ and EC 50 for NMDA, glutamate, and glycine were tested for statistical significance by two methods: 1) Concentration-response data from each oocyte were fitted to the logistic equation, and parameter estimates for E ma ⁇ and EC S0 in the presence and absence of PS were compared by unpaired, 2 -tailed t-test. 2) The normalized (to the 200 ⁇ M NMDA response) responses at each concentration from multiple oocytes were averaged to construct pooled concentration-response curves (points in Fig. 27) in the presence and absence of PS.
  • Steroids were obtained from Steraloids, Inc. (Wilton, NH) . Other compounds were obtained from Sigma (St. Louis, MO) . Steroid stocks were prepared in DMSO and diluted into recording medium (final DMSO concentration 0.5%) . Other solutions also contained 0.5% DMSO.
  • PS Pregnenolone Sulfate
  • NRla/NR2B NRla is also referred to as NR1 011
  • PS potentiated the NMDA-induced response of receptors containing NRla/NR2 Chimera (I) subunits.
  • IOOUM PS increased NRla/NR2 Chimera (I) responses by 52.69 ⁇ 3.58% in the presence of saturating concentrations of NMDA and Glycine, 300uM and 50uM, respectively (Fig. 32) .
  • residues 524 to 1482 of the NR2B were replaced with those of NR2D, PS inhibited rather than potentiated the NMDA response.
  • IOOUM PS inhibited the NRla/NR2 Chimera (II) by 43.04 ⁇ 5.44%, similar to the wild-type NRla/NR2D (45.18 ⁇ 2.62%) (Fig. 32).
  • NR2B-DChimera In the fourth chimera receptor, NR2B-DChimera (IV), the residues 727 to 892 were replaced on NR2D with residues 703 to 870 of NR2B, which contains the second extracellular domain, the fourth hydrophobic transmembrane domain and 31 residues from the C-terminal intracellular tail.
  • 100 ⁇ M PS potentiated the NR101l/NR2B-DChimera(IV) by 66.05 ⁇ 2.75%. This demonstrates that the region of residues 703 to 870 on NR2B is necessary and sufficient for the PS potentiating effect .
  • the chimeric receptors were generated by first introducing a series of silent restriction sites in both the NR2B and NR2D cDNA. The chimeric cDNAs were then constructed by replacing corresponding regions of one cDNA with that of the other.
  • the silence site mutations were generated by a method involving PCR. In brief, oligonucleotides were synthesized containing the mutation, and these oligonucleotides were used in combination with other oligonucleotides in PCR amplifications of fragments of the cDNA. The products of the PCR reactions were cut with two different restriction enzymes to generate A cassette containing the mutation. This cassette was then ligated into the cDNA cut with the same two restriction enzymes. For all of the mutations, single isolates were selected, and the entire region of the amplified cassette was sequenced to check for the mutation and insure against second-site mutations.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Microbiology (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Steroid Compounds (AREA)

Abstract

L'invention concerne un procédé d'identification d'un modulateur spécifique de sous-unité du récepteur N-méthyl-D-aspartate (NMDA). Le procédé comporte les étapes consistant à prévoir plusieurs récepteurs NMDA différents quant à leur identité de sous-unités. Les récepteurs sont mis en contact avec un ligand du site de reconnaissance de neurotransmetteurs en présence ou en l'absence d'un modulateur candidat. L'activité des récepteurs est ensuite dosée, et l'augmentation ou la diminution de l'activité d'au moins un, mais pas de tous les éléments des multiples récepteurs NMDA, en présence d'un modulateur candidat mais pas en l'absence celui-ci, indique que le modulateur candidat est un modulateur spécifique de sous-unité. L'identité de sous-unités du sous-ensemble des récepteurs NMDA détermine la spécificité de sous-unité du modulateur candidat. Diverses combinaisons de sous-unités de récepteur NMDA sont prévues.
PCT/US2000/023985 1999-08-31 2000-08-31 Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites WO2001016601A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP00959706A EP1212618B1 (fr) 1999-08-31 2000-08-31 Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites
DE60041773T DE60041773D1 (de) 1999-08-31 2000-08-31 Der effekt von steroiden auf ndma-rezeptoren beruht auf der zusammensetzung der untereinheiten
AU70981/00A AU782250B2 (en) 1999-08-31 2000-08-31 Effect of steroids on NMDA receptors depends on subunit composition
CA002383047A CA2383047A1 (fr) 1999-08-31 2000-08-31 Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15180299P 1999-08-31 1999-08-31
US60/151,802 1999-08-31

Publications (1)

Publication Number Publication Date
WO2001016601A1 true WO2001016601A1 (fr) 2001-03-08

Family

ID=22540293

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/023985 WO2001016601A1 (fr) 1999-08-31 2000-08-31 Effet de steroides sur des recepteurs nmda dependant de la composition de sous-unites

Country Status (7)

Country Link
US (1) US6623933B1 (fr)
EP (1) EP1212618B1 (fr)
AT (1) ATE425459T1 (fr)
AU (1) AU782250B2 (fr)
CA (1) CA2383047A1 (fr)
DE (1) DE60041773D1 (fr)
WO (1) WO2001016601A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1347763A1 (fr) * 2000-11-06 2003-10-01 Trustees Of Boston University Derives steroides neuroactifs et procedes d'utilisation
EP1450817A1 (fr) * 2001-08-28 2004-09-01 Monash University Utilisation de pregnane-diones ou de pregnane-diols comme agents analgesiques neuropathiques
US9687496B2 (en) 2010-01-14 2017-06-27 Asarina Pharma Ab Pharmaceutical composition comprising 3-beta-hydroxy-5-alpha-pregnan-20-one with improved storage and solubility properties
JP2017528455A (ja) * 2014-08-26 2017-09-28 ウスタフ オルガニッケ ヘミエ アー ビオヘミエ アカデミエ ヴェド ツェーエル,ヴェー.ヴェー.イー 神経保護特性を有する両親媒性化合物
WO2018103626A1 (fr) * 2016-12-05 2018-06-14 江苏恩华络康药物研发有限公司 Dérivé d'alloprégnénolone soluble dans l'eau et son utilisation

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114957445B (zh) * 2022-05-31 2023-09-19 陕西脉元生物科技有限公司 一种nmdar nr1亚基、nmdar的突变体及其构建方法和应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0606734A1 (fr) 1992-12-11 1994-07-20 Allelix Biopharmaceuticals Inc. Récepteurs du système nerveux central humain qui appartiennent à la famille NMDA-R1
WO1997003677A1 (fr) 1995-07-24 1997-02-06 Trustees Of Boston University Inhibition de l'activite des recepteurs du n-methyl-d-aspartate au moyen de derives de sulfate de pregnenolone

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0696320B1 (fr) * 1993-04-20 2002-09-25 Merck & Co., Inc. Sous-unites receptrices du n-methyl-d-aspartate humain, acides nucleiques codant pour elles, et leur utilisation
US5888996A (en) 1995-07-26 1999-03-30 Trustees Of Boston University Inhibition of NMDA receptor activity and modulation of glutamate-mediated synaptic activity
WO2001057240A2 (fr) * 2000-02-01 2001-08-09 Agy Therapeutics, Inc. Interaction du recepteur de n-methyl-d-aspartate et de la tyrosine phosphatase

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0606734A1 (fr) 1992-12-11 1994-07-20 Allelix Biopharmaceuticals Inc. Récepteurs du système nerveux central humain qui appartiennent à la famille NMDA-R1
WO1997003677A1 (fr) 1995-07-24 1997-02-06 Trustees Of Boston University Inhibition de l'activite des recepteurs du n-methyl-d-aspartate au moyen de derives de sulfate de pregnenolone
US6083941A (en) * 1995-07-24 2000-07-04 Trustees Of Boston University Inhibition of NMDA receptor activity by pregnenolone sulfate derivatives

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
"Drugs as Tools in Neurotransmitter Research", HUMANA PRESS INC., pages: 161 - 219
"Single-Channel Recording", 1995, PLENUM PRESS, pages: 341 - 356
"Transmitters and Neuromodulators: Neuroactive Steroids", 1996, CRC PRESS, pages: 23 - 36
ANSON ET AL., J. NEUROSCI., vol. 18, 1998, pages 581 - 589
BOWLBY MR, MOL PHARMACOL, vol. 43, 1993, pages 813 - 819
BREWER ET AL., BRAIN RESEARCH, vol. 494, 1989, pages 65 - 74
CHAN, P. H., STROKE, vol. 27, 1996, pages 1124 - 1129
CHOI, D. W., J NEUROBIO, vol. 23, 1992, pages 1261 - 1276
DAWSON ET AL., PROC NATL ACAD SCI USA, vol. 88, 1991, pages 6368 - 6371
DE LEAN ET AL., AM. J. PHYSIOL., vol. 235, 1978, pages E97 - E102
FARB DH; GIBBS TT: "CNS neurotransmitters and neuromodulators: neuroactive steroids", 1996, CRC PRESS, article "Steroids as modulators of amino acid receptor function", pages: 23 - 36
G6MEZ-PINILLA ET AL., EXP NEUROL, vol. 104, 1989, pages 118 - 124
GREENAMYRE ET AL., ARCH NEUROL, vol. 48, 1991, pages 977 - 981
GREENAMYRE, J.T., NEUROBIOL AGING, vol. 12, 1991, pages 334 - 336
HOLLMANN; HEINEMANN, ANNU. REV. NEUROSCI., vol. 17, 1994, pages 31 - 108
HONORE ET AL., SCIENCE, vol. 84, 1988, pages 8215 - 8219
IRWIN ET AL., J. PHARMACOL. EXP. THER., vol. 271, 1994, pages 677 - 682
IRWIN ET AL.: "Pregnenolone sulfate augments NMDA receptor mediated increases in intracellular CA2+ in cultured rat hippocampal neurons", NEUROSCI. LETT., vol. 141, 1992, pages 30 - 34, XP002934866 *
LAURIE ET AL., EUR. J. PHARM., vol. 268, 1994, pages 335 - 345
LAURIE ET AL.: "Ligand affinities at recombinant N-methyl-D-aspartate receptors depend on subunit composition", EUR J. PHARM., vol. 268, 1994, pages 335 - 345, XP002934864 *
LEONARD; KELSO, NEURON, vol. 4, 1990, pages 53 - 60
MCBAIN ET AL.: "N-methyl-D-aspartic acid receptor structure and function", PHYSIOL. REV., vol. 74, no. 3, July 1994 (1994-07-01), pages 723 - 760, XP002934863 *
MUNSON; RODBARD, ANAL. BIOCHEM., vol. 107, 1980, pages 220 - 239
NAKANISHI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 89, 1992, pages 8552 - 8556
PARK-CHUNG ET AL., MOL PHARMACOL, vol. 46, 1994, pages 146 - 150
PARK-CHUNG ET AL., MOL PHARMACOL, vol. 52, 1997, pages 1113 - 1123
PARK-CHUNG ET AL., MOL. PHARMACOL., vol. 46, 1994, pages 146 - 150
PARK-CHUNG ET AL., MOL. PHARMACOL., vol. 52, 1997, pages 1113 - 1123
PARK-CHUNG ET AL.: "Distinct sites for inverse modulation of N-methyl-D-aspartate receptors by sulfated steroids", MOL. PHARM., vol. 52, 1997, pages 1113 - 1123, XP002934865 *
PIKE ET AL., J NEUROSCI, vol. 13, 1993, pages 1676 - 1687
ROTHMAN ET AL., ANN NEUROLOGY, vol. 19, 1986, pages 105 - 111
See also references of EP1212618A4
SOLOVIEV; BARNARD, J. MOL. BID., vol. 273, 1997, pages 14 - 18
WAFFORD ET AL., MOL. PHARMACOL., vol. 47, 1995, pages 374 - 380
WEAVER ET AL., BRAIN RES, vol. 761, 1997, pages 338 - 341
WEAVER ET AL., PROC NATL ACAD SCI USA, vol. 94, 1997, pages 10450 - 10454
WEAVER ET AL.: "17beta-Estradiol protects against NMDA-induced excitotoxicity by direct inhibition of NMDA receptors", BRAIN RES., vol. 761, 1997, pages 338 - 341, XP002934867 *
WISDEN; AND SEEBURG, J. NEUROSCI., vol. 13, 1993, pages 3582 - 3598
WU ET AL., MOL PHARMACOL, vol. 40, 1991, pages 333 - 336
WU ET AL., MOL. PHARMACOL., vol. 40, 1991, pages 333 - 336
YAGHOUBI ET AL., BRAIN RES., vol. 803, 1998, pages 153 - 160
YAGHOUBI ET AL.: "20", SOC. NEUROSCI., 1995, pages 1109

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1347763A1 (fr) * 2000-11-06 2003-10-01 Trustees Of Boston University Derives steroides neuroactifs et procedes d'utilisation
EP1347763A4 (fr) * 2000-11-06 2005-10-12 Univ Boston Derives steroides neuroactifs et procedes d'utilisation
EP1450817A1 (fr) * 2001-08-28 2004-09-01 Monash University Utilisation de pregnane-diones ou de pregnane-diols comme agents analgesiques neuropathiques
JP2005504776A (ja) * 2001-08-28 2005-02-17 モナシュ ユニバーシティ 神経因性鎮痛薬としてのプレグナン−ジオンまたはジオールの使用
EP1450817A4 (fr) * 2001-08-28 2009-02-25 Univ Monash Utilisation de pregnane-diones ou de pregnane-diols comme agents analgesiques neuropathiques
US9687496B2 (en) 2010-01-14 2017-06-27 Asarina Pharma Ab Pharmaceutical composition comprising 3-beta-hydroxy-5-alpha-pregnan-20-one with improved storage and solubility properties
US11534446B2 (en) 2010-01-14 2022-12-27 Asarina Pharma Ab Pharmaceutical composition comprising 3-beta-hydroxy-5-alpha-pregnan-20-one with improved storage and solubility properties
JP2017528455A (ja) * 2014-08-26 2017-09-28 ウスタフ オルガニッケ ヘミエ アー ビオヘミエ アカデミエ ヴェド ツェーエル,ヴェー.ヴェー.イー 神経保護特性を有する両親媒性化合物
WO2018103626A1 (fr) * 2016-12-05 2018-06-14 江苏恩华络康药物研发有限公司 Dérivé d'alloprégnénolone soluble dans l'eau et son utilisation

Also Published As

Publication number Publication date
AU7098100A (en) 2001-03-26
ATE425459T1 (de) 2009-03-15
CA2383047A1 (fr) 2001-03-08
EP1212618A1 (fr) 2002-06-12
AU782250B2 (en) 2005-07-14
EP1212618B1 (fr) 2009-03-11
DE60041773D1 (de) 2009-04-23
EP1212618A4 (fr) 2005-04-06
US6623933B1 (en) 2003-09-23

Similar Documents

Publication Publication Date Title
Maurice et al. Neuro (active) steroids actions at the neuromodulatory sigma1 (σ1) receptor: biochemical and physiological evidences, consequences in neuroprotection
Jaber et al. Dopamine receptors and brain function
Lambert et al. Neurosteroid modulation of native and recombinant GABA A receptors
Akk et al. Mechanisms of neurosteroid interactions with GABAA receptors
Korinek et al. Neurosteroid modulation of N-methyl-D-aspartate receptors: molecular mechanism and behavioral effects
Paul et al. Neuroactive steroids
Gu et al. Rapid action of 17β-estradiol on kainate-induced currents in hippocampal neurons lacking intracellular estrogen receptors
Xu et al. Interaction of picrotoxin with GABAA receptor channel-lining residues probed in cysteine mutants
Krasowski et al. General anaesthetic actions on ligand-gated ion channels
Kumar et al. Nestorone® as a novel progestin for nonoral contraception: structure-activity relationships and brain metabolism studies
Arias Binding sites for exogenous and endogenous non-competitive inhibitors of the nicotinic acetylcholine receptor
Maurice et al. Neuroactive neurosteroids as endogenous effectors for the sigma1 (σ1) receptor: pharmacological evidence and therapeutic opportunities
Lambert et al. Steroid modulation of the GABAA receptor complex: electrophysiological studies
Lan et al. Identification and characterization of a pregnane steroid recognition site that is functionally coupled to an expressed GABA A receptor
DE60013417T2 (de) Verfahren zum screenen von fxr-rezeptormodulatoren
Zhou et al. PMP22 regulates cholesterol trafficking and ABCA1-mediated cholesterol efflux
AU2657292A (en) Novel gabaa receptor with steroid binding sites
Barrantes Structural‐functional correlates of the nicotinic acetylcholine receptor and its lipid microenvironment
Lafont et al. 20-Hydroxyecdysone activates the protective arm of the RAAS via the MAS receptor
Li et al. Detecting the presence of hippocampus membrane androgen receptors in male SAMP8 mice and their induced synaptic plasticity
US6063582A (en) Methods of screening for compounds that interact with human P2u2 purinergic receptor
US6623933B1 (en) Methods for identifying a subunit specific modulator of N-methyl-D-asparate receptor
Hansen et al. Pharmacological characterization of ligands at recombinant NMDA receptor subtypes by electrophysiological recordings and intracellular calcium measurements
US20040204490A1 (en) Effect of steroids on NMDA receptors depends on subunit composition
Ljungstrom et al. Functional coupling between heterologously expressed dopamine D 2 receptors and KCNQ channels

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2383047

Country of ref document: CA

Ref document number: 70981/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2000959706

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2000959706

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: JP

WWG Wipo information: grant in national office

Ref document number: 70981/00

Country of ref document: AU